1
|
Constanthin PE, Contestabile A, Petrenko V, Quairiaux C, Salmon P, Hüppi PS, Kiss JZ. Endogenous erythropoietin signaling regulates migration and laminar positioning of upper-layer neurons in the developing neocortex. Development 2020; 147:dev190249. [PMID: 32764029 PMCID: PMC7561482 DOI: 10.1242/dev.190249] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/24/2020] [Indexed: 01/24/2023]
Abstract
Erythropoietin (EPO), the hypoxia-inducible hematopoietic hormone, has well-established neuroprotective/neurotrophic roles in the developing central nervous system and the therapeutic potential of EPO has been widely explored in clinical studies for the treatment of perinatal hypoxic brain lesion, as well as prematurity. Here, we reveal that both EPO and Epo receptor (EPOR) are expressed in the developing rat somatosensory cortex during radial migration and laminar positioning of granular and supragranular neurons. Experimental deregulation of EPO signaling using genetic approaches results in aberrant migration, as well as permanent neuronal misplacement leading to abnormal network activity and protracted sensory behavioral deficits. We identify ERK as the downstream effector of the EPO signaling pathway for neuronal migration. These findings reveal a crucial role for endogenous EPO signaling in neuronal migration, and offer important insights for understanding how the temporary deregulation of EPO could result in migration defects that lead to abnormal behavior in the adult.
Collapse
Affiliation(s)
- Paul E Constanthin
- Department of Fundamental Neurosciences, University Medical Center, University of Geneva, 1201 Geneva, Switzerland
| | - Alessandro Contestabile
- Department of Fundamental Neurosciences, University Medical Center, University of Geneva, 1201 Geneva, Switzerland
| | - Volodymyr Petrenko
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, 1201 Geneva, Switzerland
- Department of Cell Physiology and Metabolism; Diabetes Center, Faculty of Medicine, University of Geneva; Institute of Genetics and Genomics in Geneva (iGE3), 1201 Geneva, Switzerland
| | - Charles Quairiaux
- Department of Fundamental Neurosciences, University Medical Center, University of Geneva, 1201 Geneva, Switzerland
| | - Patrick Salmon
- Department of Fundamental Neurosciences, University Medical Center, University of Geneva, 1201 Geneva, Switzerland
| | - Petra S Hüppi
- Department of Pediatrics, Faculty of Medicine, University Hospital of Geneva, 1201 Geneva, Switzerland
| | - Jozsef Z Kiss
- Department of Fundamental Neurosciences, University Medical Center, University of Geneva, 1201 Geneva, Switzerland
| |
Collapse
|
2
|
Guha Majumdar A, Subramanian M. Hydroxychavicol from Piper betle induces apoptosis, cell cycle arrest, and inhibits epithelial-mesenchymal transition in pancreatic cancer cells. Biochem Pharmacol 2019; 166:274-291. [PMID: 31154000 DOI: 10.1016/j.bcp.2019.05.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/28/2019] [Indexed: 12/30/2022]
Abstract
Pancreatic cancer is a major cause of cancer-related mortality around the world. Currently, options for diagnosis and treatment are extremely limited, which culminates in a very high mortality rate. Intensive research spanning more than four decades has met several roadblocks in terms of improvement in overall survival. In this study, we have evaluated the effect of Hydroxychavicol (HC), a naturally occurring and abundantly isolatable allylarene from Piper betle leaves on pancreatic cancer cells. Our investigation reveals that HC inhibits proliferation and epithelial-mesenchymal transition (EMT) in pancreatic cancer cells. HC induces DNA damage, as evidenced by γ-H2AX, 53BP1 induction and comet assay, which further results in mitotic catastrophe and apoptosis. The apoptosis induced by HC is JNK pathway-dependent and caspase-mediated. HC also inhibits migration and invasion of pancreatic cancer cells via a generalized repression of genes involved in EMT. A quantitative real time PCR-based array revealed at least 14 different genes to be differentially expressed upon HC treatment in pancreatic cancer cells. These results show significant potential of HC as an anticancer agent against pancreatic cancer.
Collapse
Affiliation(s)
- Ananda Guha Majumdar
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| | - Mahesh Subramanian
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India.
| |
Collapse
|
3
|
Vasculogenic Mimicry Formation Is Associated with Erythropoietin Expression but Not with Erythropoietin Receptor Expression in Cervical Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1934195. [PMID: 30915348 PMCID: PMC6409067 DOI: 10.1155/2019/1934195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/30/2018] [Accepted: 01/10/2019] [Indexed: 12/24/2022]
Abstract
Background Vasculogenic mimicry (VM), as an endothelium-independent cancer microcirculation, has been observed in many malignancies including cervical cancer. Erythropoietin (EPO) and erythropoietin receptor (EPO-R) could produce an angiogenic effect to promote cervical squamous cell carcinoma (CSCC) progression. However, the association between VM formation and EPO/EPO-R expression in CSCC is poorly explored. Methods Seventy-six paraffin-embedded CSCC samples, 25 high-grade squamous intraepithelial lesion (HSIL) samples, 20 low-grade squamous intraepithelial lesion (LSIL) samples, and 20 normal cervix samples were collected. Immunohistochemistry SP method was performed to detect EPO/EPO-R expression and CD31/periodic acid-Schiff (PAS) double staining was performed to detect VM formation. The associations of EPO/EPO-R and VM with clinicopathological parameters of CSCC were analyzed. The associations between VM formation and EPO/EPO-R expression were also analyzed. Results The positive expression rates of EPO and EPO-R were gradually increasing along the progression of normal cervix-LSIL-HSIL-CSCC sequence (P<0.05). EPO and EPO-R expression were not significantly associated with clinicopathological parameters of CSCC patients (P>0.05). VM was significantly associated with FIGO stage, lymphovascular space involvement, and lymph node metastasis (P<0.05). VM was positively associated with EPO expression (r=0.284, P<0.05) but was not associated with EPO-R expression (P>0.05). Conclusion These data suggest that increased EPO/EPO-R expression may play an important role in cervical carcinogenesis. EPO overexpression may promote VM formation in CSCC.
Collapse
|
4
|
Li Y, Zhong W, Zhu M, Hu S, Su X. Nodal regulates bladder cancer cell migration and invasion via the ALK/Smad signaling pathway. Onco Targets Ther 2018; 11:6589-6597. [PMID: 30323631 PMCID: PMC6178944 DOI: 10.2147/ott.s177514] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Bladder cancer is the most common malignant tumor of the urinary tract. We aimed to explore the biological role and molecular mechanism of Nodal in bladder cancer. Materials and methods The expression of Nodal in bladder cancer tissues and cells was determined by quantitative real-time polymerase chain reaction. The effect of silencing of Nodal on cell proliferation, clone formation, and migration and invasion was evaluated by MTT cell proliferation assay, colony formation, and transwell assays, respectively. Western blot analysis was employed to detect the expression of proliferation- and invasion-related proteins and proteins involved in ALK/Smad signaling. Results We found that the expression of Nodal was significantly increased in bladder cancer tissues and cell lines. Downregulation of Nodal effectively weakened cell proliferation, clone formation, and cell migration and invasion abilities. The protein expression levels of CDC6, E-cadherin, MMP-2, and MMP-9 were also altered by downregulation of Nodal. Knockdown of Nodal also blocked the expression of ALK4, ALK7, Smad2, and Smad4, which are involved in ALK/Smad signaling. Additionally, the ALK4/7 receptor blocker SB431542 reversed the promotive effects of Nodal overexpression on bladder cancer cell proliferation, migration, and invasion. Conclusion Our study indicated that Nodal functions as an oncogene by regulating cell proliferation, migration, and invasion in bladder cancer via the ALK/Smad signaling pathway, thereby providing novel insights into its role in bladder cancer treatment.
Collapse
Affiliation(s)
- Youkong Li
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou 434020, People's Republic of China,
| | - Wen Zhong
- Department of Endocrine, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou 434020, People's Republic of China
| | - Min Zhu
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou 434020, People's Republic of China,
| | - Shengguo Hu
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou 434020, People's Republic of China,
| | - Xiaokang Su
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou 434020, People's Republic of China,
| |
Collapse
|
5
|
Ordoñez-Moreno A, Rodriguez-Monterrosas C, Cortes-Reynosa P, Perez-Carreon JI, Perez Salazar E. Erythropoietin Induces an Epithelial to Mesenchymal Transition-Like Process in Mammary Epithelial Cells MCF10A. J Cell Biochem 2017; 118:2983-2992. [PMID: 28247960 DOI: 10.1002/jcb.25959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/27/2017] [Indexed: 11/06/2022]
Abstract
Anemia is associated with chemotherapy treatment in cancer patients. Erythropoietin (EPO) has been used to treat anemia of cancer patients, because it stimulates erythropoiesis. However, treatment of breast cancer patients with EPO has been associated with poor prognosis and decrease of survival. Epithelial to mesenchymal transition (EMT) is a process by which epithelial cells are transdifferentiated to a mesenchymal state. It has been implicated in tumor progression, because epithelial cells acquire the capacity to execute the multiple steps of invasion/metastasis process. However, the role of EPO on EMT process in human mammary epithelial cells has not been studied. In the present study, we demonstrate that EPO promotes a decrease of E-cadherin expression, an increase of N-cadherin, vimentin, and Snail2 expression, activation of FAK and Src kinases and an increase of MMP-2 and MMP-9 secretions. Moreover, EPO induces an increase of NFκB DNA binding activity, an increase of binding of p50 and p65 NFκB subunits to Snail1 promoter, migration, and invasion in mammary non-tumorigenic epithelial cells MCF10A. In summary, these findings demonstrate, for the first time, that EPO induces an EMT-like process in mammary non-tumorigenic epithelial cells. J. Cell. Biochem. 118: 2983-2992, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | - Pedro Cortes-Reynosa
- Departamento de Biología Celular, Cinvestav-IPN, Av. IPN # 2508, San Pedro Zacatenco, Mexico
| | | | - Eduardo Perez Salazar
- Departamento de Biología Celular, Cinvestav-IPN, Av. IPN # 2508, San Pedro Zacatenco, Mexico
| |
Collapse
|
6
|
Du E, Zhang C, Qin Z, Yang K, Li C, Wang A, Zhang Z, Xu Y. Low expression of TMEM67 is a critical predictor of poor prognosis in human urothelial carcinoma of the bladder. Urol Oncol 2017; 35:152.e7-152.e12. [PMID: 28161324 DOI: 10.1016/j.urolonc.2016.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/15/2016] [Accepted: 10/19/2016] [Indexed: 11/29/2022]
Abstract
OBJECTIVES The aim of the study was to evaluate the expression of TMEM67 in urothelial carcinoma of the bladder (UCB) tissues and to determine the potential relevance between the expression of TMEM67 and prognosis of UCB. MATERIAL AND METHODS In this study, the expression of TMEM67 mRNA was performed by quantitative real-time PCR in 80 UCB and 54 noncancerous tissues. The expression of TMEM67 protein was identified by immunohistochemistry and western blotting. Chi-square test was conducted to verify the relevance between the expression of TMEM67 and clinical parameters. Kaplan-Meier survival analysis was demonstrated between high or low expression level of TMEM67 mRNA and recurrence-free survival probability. Cox regression analysis was conducted to evaluate the relevance between the expression of TMEM67 and the prognosis in UCB. RESULTS Low expression of TMEM67 mRNA and protein was detected in most of UCB tissues using quantitative real-time polymerase chain reaction and western blotting, compared with noncancerous tissues. Low expressions of TMEM67 were associated with TNM stage, grade, and lymph node metastasis (P<0.05). Kaplan-Meier analysis showed that the low expression of TMEM67 mRNA had significantly shorter recurrence-free survival probability (P = 0.018). Cox regression analysis confirmed that low expression of TMEM67 mRNA predicted poor prognosis of patients with UCB (HR = 2.950, P = 0.029, 95% CI: 1.116-7.796). CONCLUSIONS TMEM67 expression is low in UCB tissues, and the TMEM67 low expression predicted poor prognosis of patients with UCB.
Collapse
Affiliation(s)
- E Du
- Tianjin Institute of Urology, The 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Changwen Zhang
- Tianjin Institute of Urology, The 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Zhenbang Qin
- Tianjin Institute of Urology, The 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Kuo Yang
- Tianjin Institute of Urology, The 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Changying Li
- Tianjin Institute of Urology, The 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Aixiang Wang
- Tianjin Institute of Urology, The 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Zhihong Zhang
- Tianjin Institute of Urology, The 2nd Hospital of Tianjin Medical University, Tianjin, China.
| | - Yong Xu
- Tianjin Institute of Urology, The 2nd Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
7
|
Luo W, Hu L, Li W, Xu G, Xu L, Zhang C, Wang F. Epo inhibits the fibrosis and migration of Müller glial cells induced by TGF-β and high glucose. Graefes Arch Clin Exp Ophthalmol 2016; 254:881-90. [DOI: 10.1007/s00417-016-3290-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 01/06/2016] [Accepted: 02/08/2016] [Indexed: 12/11/2022] Open
|
8
|
Yang J, Yuan D, Li J, Zheng S, Wang B. miR-186 downregulates protein phosphatase PPM1B in bladder cancer and mediates G1-S phase transition. Tumour Biol 2015; 37:4331-41. [DOI: 10.1007/s13277-015-4117-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 09/20/2015] [Indexed: 02/05/2023] Open
|
9
|
Xiao J, Liu L, Zhong Z, Xiao C, Zhang J. Mangiferin regulates proliferation and apoptosis in glioma cells by induction of microRNA-15b and inhibition of MMP-9 expression. Oncol Rep 2015; 33:2815-20. [PMID: 25901555 DOI: 10.3892/or.2015.3919] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/02/2015] [Indexed: 11/05/2022] Open
Abstract
Mangiferin, a flavonoid extracted from the leaves of the Anacardiaceae plant, the mango tree, has physiological activity and pharmacological effects in many aspects. The present study aimed to clarify the effect of mangiferin on proliferation and apoptosis of glioma cells and the mechanism of these curative effects of mangiferin. In this experiment, we detected the proliferation using 3-(4,5-dimethylthylthiazol-2-yl)-2,5 diphenyltetrazolium bromide (MTT) assay. Then, cell apoptosis of U87 glioma cells was measured with the Annexin V-FITC/propidium iodide (PI) apoptosis detection kit, DAPI staining assay and the caspase-3 and caspase-9 activity assay kit. Next, quantitative real-time PCR and gelatin zymography were used to analyze the expression of microRNA-15b (miR-15b) and matrix metalloproteinase-9 (MMP-9), respectively. MMP-9 agonist, miR-15b mimics and anti-miR-15b mimics were added to the U87 glioma cells for elucidating the mechanisms involved in the curative effects of mangiferin. In the present study, mangiferin notably restrained the proliferation and increased the apoptosis of the U87 glioma cells. Meanwhile, mangiferin specifically promoted the expression of miR-15b and suppressed the level of MMP-9 in the U87 glioma cells. miR-15b regulated the expression of MMP-9 in the U87 glioma cells. MMP-9 agonist and anti-miR‑15b reduced the curative effects of mangiferin in the U87 glioma cells. In summary, mangiferin regulates proliferation and apoptosis in glioma cells by induction of miR-15b and inhibition of MMP-9 expression.
Collapse
Affiliation(s)
- Jinsong Xiao
- Division of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Li Liu
- Division of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zian Zhong
- Division of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Cheng Xiao
- Division of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Junjian Zhang
- Division of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
10
|
Ong Q, Guo S, Zhang K, Cui B. U0126 protects cells against oxidative stress independent of its function as a MEK inhibitor. ACS Chem Neurosci 2015; 6:130-7. [PMID: 25544156 PMCID: PMC4304487 DOI: 10.1021/cn500288n] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
![]()
U0126 is a potent
and selective inhibitor of MEK1 and MEK2 kinases.
It has been widely used as an inhibitor for the Ras/Raf/MEK/ERK signaling
pathway with over 5000 references on the NCBI PubMed database. In
particular, U0126 has been used in a number of studies to show that
inhibition of the Raf/MEK/ERK pathway protects neuronal cells against
oxidative stress. Here, we report that U0126 can function as an antioxidant
that protects PC12 cells against a number of different oxidative-stress
inducers. This protective effect of U0126 is independent of its function
as a MEK inhibitor, as several other MEK inhibitors failed to show
similar protective effects. U0126 reduces reactive oxygen species
(ROS) in cells. We further demonstrate that U0126 is a direct ROS
scavenger in vitro, and the oxidation products of U0126 exhibit fluorescence.
Our finding that U0126 is a strong antioxidant signals caution for
its future usage as a MEK inhibitor and for interpreting some previous
results.
Collapse
Affiliation(s)
- Qunxiang Ong
- Department
of Chemistry, Stanford University, 380 Roth Way, Palo Alto, California 94305, United States
| | - Shunling Guo
- Department
of Chemistry, Stanford University, 380 Roth Way, Palo Alto, California 94305, United States
| | - Kai Zhang
- Department
of Chemistry, Stanford University, 380 Roth Way, Palo Alto, California 94305, United States
- Department
of Biochemistry, University of Illinois at Urbana−Champaign, 600 South Mathews, Urbana, Illinois 61801, United States
| | - Bianxiao Cui
- Department
of Chemistry, Stanford University, 380 Roth Way, Palo Alto, California 94305, United States
| |
Collapse
|